Previous Article in Journal
Facilitating and Hindering Factors in the Implementation of a Care Transition Strategy: Mixed Methods Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Akkermansia muciniphila in Cardiometabolic Medicine: Mechanisms, Clinical Studies, and Therapeutic Outlook

by
Alireza FakhriRavari
1,* and
Minh Hien Chau Nguyen
2
1
Department of Pharmacy Practice, School of Pharmacy, Loma Linda University, Loma Linda, CA 92350, USA
2
CVS Health, Rancho Cucamonga, CA 91730, USA
*
Author to whom correspondence should be addressed.
Gastrointest. Disord. 2025, 7(4), 72; https://doi.org/10.3390/gidisord7040072 (registering DOI)
Submission received: 28 September 2025 / Revised: 25 October 2025 / Accepted: 7 November 2025 / Published: 9 November 2025

Abstract

Akkermansia muciniphila—a mucus-resident commensal—has emerged as a promising target at the interface of metabolism, barrier function, and immunity. Observational human studies link higher intestinal abundance of A. muciniphila with healthier adiposity and glycemic profiles, while preclinical experiments demonstrate causal benefits on adiposity, insulin resistance, gut-barrier integrity, and inflammatory tone. These effects are attributed to mucus-layer reinforcement, reduced intestinal permeability and endotoxemia, production of short-chain fatty acids, and host signaling by defined bacterial components. In a randomized proof-of-concept trial in overweight/obese insulin-resistant adults, pasteurized A. muciniphila was safe and well-tolerated and improved insulin sensitivity and total cholesterol versus placebo; live cells showed directionally favorable but non-significant trends. A separate multicenter randomized trial of a five-strain consortium that included A. muciniphila improved post-prandial glucose and HbA1c in type 2 diabetes, supporting translational potential while underscoring the need for strain-resolved studies. Evidence for liver and cardiovascular benefits is strong in animals (e.g., MASLD and atherosclerosis models) but remains preliminary in humans. Inter-individual response heterogeneity—potentially influenced by baseline Akkermansia levels and gut-barrier status—highlights the value of personalized, microbiome-guided approaches. Larger, longer clinical studies are now warranted to define optimal dosing and formulation (live vs. pasteurized), durability, safety across populations, and impacts on hard outcomes (clinically meaningful weight change, glycemic endpoints, and cardiometabolic events). Overall, A. muciniphila represents a promising microbial adjunct for metabolic health with a plausible path from postbiotic concepts to clinical application, pending confirmatory trials.

1. Introduction

Akkermansia muciniphila is a Gram-negative, non-spore-forming, non-motile, strictly anaerobic bacterium that resides in the mucus layer of the gut [1,2,3]. First isolated in 2004 by Derrien and colleagues (strain MucT), this oval-shaped bacterium belongs to the phylum Verrucomicrobiota (formerly Verrucomicrobia) and has since been recognized as a key member of the healthy human gut microbiota [3]. A. muciniphila specializes in degrading mucin, the glycoprotein component of mucus, and in turn produces short-chain fatty acids (SCFAs) like acetate and propionate that nourish other beneficial gut bacteria [3]. Its presence is associated with a thicker intestinal mucus layer and enhanced gut barrier function in animal studies [4]. Over the past decade, accumulating evidence has linked A. muciniphila to various aspects of human health, sparking interest in its potential as a “next-generation probiotic” [5].
Numerous human studies have observed an inverse correlation between A. muciniphila abundance and metabolic disorders. Individuals with obesity, type 2 diabetes, or metabolic syndrome often have reduced levels of A. muciniphila in their gut, whereas lean and healthy individuals show higher levels [6,7,8,9,10,11,12]. Similar patterns are seen in animal models: mice with diet-induced obesity or diabetic traits tend to have diminished A. muciniphila populations [13]. These observations suggest that A. muciniphila may play a protective role against metabolic dysfunctions. Indeed, A. muciniphila has been proposed as a promising therapeutic microbe for alleviating obesity, diabetes, and related comorbidities [14]. Beyond metabolic health, correlations extend to gut inflammatory conditions; lower levels of A. muciniphila have been reported in patients with inflammatory bowel disease (IBD) and other chronic inflammatory states [15]. Such observations imply that boosting A. muciniphila might benefit not only metabolism but also gut health and immune balance.
Importantly, this literature differentiates between probiotics and postbiotics. In the International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus, probiotics are “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host.” By contrast, postbiotics are defined as “preparations of inanimate microorganisms and/or their components that confer a health benefit.” The latter category explicitly includes heat-treated (pasteurized) cells and their cellular components, which is relevant to A. muciniphila because pasteurization preserves key bioactive structures [16].
Prompted by these correlations, researchers have investigated whether supplementing the diet with A. muciniphila can improve health outcomes. Early experimental studies in rodents provided proof-of-concept: administration of A. muciniphila to mice on a high-fat diet curbed weight gain, improved metabolic parameters, and reduced inflammation [13]. These findings were pivotal in demonstrating a causal link between A. muciniphila and host health, fueling the idea that this bacterium could be harnessed as a probiotic. Importantly, A. muciniphila’s mode of action appears to involve strengthening the gut’s defensive barrier and modulating host metabolic and immune pathways [17]. Until recently, A. muciniphila had never been given as a supplement to humans, but pioneering clinical trials have now begun to test its safety and efficacy [18]. Early human data show promise in metabolic health improvement, aligning with the animal studies.
In light of these developments, A. muciniphila has moved from being merely a gut microbe of interest to an active ingredient in new probiotic (or “postbiotic”) formulations. This review provides a clinical overview of the role of A. muciniphila supplementation on health. We examine its impacts on obesity, diabetes, gut health, and inflammation, among other conditions. We discuss the mechanisms by which A. muciniphila exerts its effects, the results of preclinical and clinical studies, and the emergence of commercially available A. muciniphila supplements. By surveying the current literature, we aim to clarify how supplementing with Akkermansia muciniphila might be leveraged to improve human health and what considerations accompany its use.

2. Results

2.1. Impact on Obesity and Metabolic Syndrome

One of the most extensively studied areas for A. muciniphila supplementation is obesity and the cluster of conditions known as metabolic syndrome. In animal models, the introduction of A. muciniphila has consistently yielded anti-obesity effects. Research by Everard and colleagues reported that adding A. muciniphila to the diet of mice fed a high-fat, obesogenic diet led to a significant reduction in body fat accumulation and metabolic disturbances [13]. In these mice, A. muciniphila treatment attenuated weight gain without altering food intake, suggesting improved metabolic efficiency rather than caloric intake reduction. Notably, markers of metabolic health improved: treated mice showed reduced adipose tissue inflammation and enhanced insulin sensitivity compared to untreated obese mice. The presence of A. muciniphila was also associated with higher levels of endocannabinoids, molecules that help regulate glucose and energy homeostasis, and a strengthening of the intestinal barrier. These changes collectively contributed to better metabolic outcomes. Early studies indicated that live, active bacteria were required for these benefits, as heat-killed bacteria initially showed no effect in the 2013 mouse experiments [13,19]. This finding pointed to the importance of A. muciniphila’s metabolic activity or structural integrity in conferring benefits (a point later revisited in human studies with pasteurized bacteria).
Further rodent studies reinforced the anti-obesity impact of A. muciniphila [20]. In diet-induced obese mice, daily oral gavage of A. muciniphila for several weeks significantly alleviated weight gain and reduced fat mass accumulation. Treated mice had improvements in metabolic syndrome indicators, including lower fasting blood glucose and insulin levels, improved insulin sensitivity, and better lipid profiles. For example, one study found A. muciniphila administration led to a reduction in plasma cholesterol and triglycerides, accompanied by decreased adipose tissue inflammation [21]. Mechanistically, the anti-obesity effect in mice has been linked to A. muciniphila’s ability to enhance the gut barrier and reduce systemic inflammation. Treated obese mice exhibit increased expression of intestinal tight junction proteins and a thicker mucus layer, which together reduce gut permeability and endotoxin leakage [22,23]. By preventing the passage of pro-inflammatory bacterial components like lipopolysaccharide (LPS) into circulation, A. muciniphila lowers the chronic low-grade inflammation that characterizes obesity. Indeed, mice receiving A. muciniphila show reduced levels of inflammatory cytokines in fat tissue and increased markers of anti-inflammatory regulatory T cells, reflecting an attenuation of obesity-related inflammation [24].
Crucially, the benefits of A. muciniphila supplementation on obesity are not limited to rodents. Plovier and colleagues first addressed key translational hurdles by cultivating A. muciniphila in a fully synthetic, animal-component-free medium compatible with human administration; unexpectedly, they also showed that pasteurization enhanced efficacy in obese/diabetic mice, yielding larger improvements in fat mass, insulin resistance, and dyslipidemia than the live bacterium. Plovier and colleagues published the first-in-human, proof-of-concept study (NCT02637115) of A. muciniphila MucT in 2017 [25]. In a preliminary analysis, they randomized 20 adults (5 per group) to placebo, live A. muciniphila (109 or 1010 cells/day), or pasteurized A. muciniphila (1010 cells/day) for two weeks and observed no safety signal: standard safety parameters (inflammation, hematology, kidney, liver, muscle) were unchanged and adverse-event frequency was similar across groups (a few cases of borborygmi with live bacteria, not significantly different from other arms).
The full results of this pilot were published by Depommier and colleagues in 2019 and included overweight and obese, insulin-resistant adults [18]. In this randomized, double-blind, single-center, placebo-controlled trial (40 randomized; 32 completed), participants received either live A. muciniphila (1010 bacteria per day), pasteurized A. muciniphila (1010 bacteria per day), or a placebo daily for three months. The primary outcomes were safety, tolerability, and metabolic parameters. Supplementation (live or pasteurized) was safe and well-tolerated, with adverse-event rates comparable to placebo. In terms of efficacy, while live A. muciniphila trended toward improvements (insulin sensitivity +42.4% vs. placebo; insulinemia −12.0%; plasma total cholesterol −6.7%; body weight −0.81 kg; fat mass −0.79 kg; hip circumference −1.05 cm; changes generally not statistically significant), it was the pasteurized A. muciniphila that achieved significant benefits. The pasteurized bacteria supplementation resulted in increased insulin sensitivity (+28.6%; p = 0.002), decreased insulinemia (−34.1%; p = 0.006), and lower plasma total cholesterol (−8.7%; p = 0.02) versus placebo. Reductions in body weight (−2.27 kg over 3 months; p = 0.09), fat mass (−1.37 kg; p = 0.09), and hip circumference (−2.63 cm; p = 0.09) showed clear trends without reaching statistical significance. While these anthropometric changes were moderate, they occurred without any other lifestyle intervention, underscoring the potential of A. muciniphila to aid weight management. Overall, both animal and human findings support that A. muciniphila supplementation can beneficially influence weight and adiposity, particularly in the context of a high-fat diet or metabolic syndrome. The consistent observation is that A. muciniphila helps create a metabolic environment less prone to fat storage and inflammation, thereby counteracting obesity’s drivers.
In addition, more recent human trials extend these findings. First, a multicenter RCT in adults with overweight/obese type 2 diabetes reported that supplementation with A. muciniphila (strain AKK-WST01) improved clinical endpoints—but predominantly in those with low baseline intestinal A. muciniphila, who showed higher colonization alongside significant reductions in body weight, fat mass, and HbA1c; participants with high baseline Akkermansia exhibited poor colonization and no significant benefit (NCT04797442) [26]. These data suggest efficacy may depend on baseline gut levels, pointing to a microbiota-guided personalization strategy. Second, an 8-week randomized, double-blind trial testing postbiotic (heat-inactivated) A. muciniphila delivered in yogurt in adults with overweight/obesity (n = 66) found significant between-group reductions in waist circumference, waist-to-height ratio, body fat percentage, and AST in the Akkermansia-yogurt arm versus control, while a comparator yogurt fortified with Lactobacillus rhamnosus postbiotic showed no between-group benefits [27].
Overall, the results across multiple studies indicate that supplementing with Akkermansia muciniphila produces beneficial outcomes in obesity and metabolic syndrome. In obese hosts, it promotes a leaner phenotype, improves metabolic markers (glucose, lipids), and mitigates tissue inflammation. These effects have been demonstrated robustly in animal models and are now being observed to some extent in humans [18]. The ability of A. muciniphila to fortify the gut barrier and reduce systemic inflammatory tone appears central to its impact on obesity. Notably, its efficacy in reducing weight gain and metabolic dysfunction positions A. muciniphila as a unique probiotic candidate targeting obesity—a condition traditionally managed with diet, exercise, and medication. The promising human data (albeit from a small trial), together with baseline-dependent efficacy in type 2 diabetes and positive signals from a postbiotic yogurt delivery, further suggest that A. muciniphila supplementation could become a novel tool in weight management and metabolic health, pending confirmation from larger studies. A side-by-side summary of human clinical studies of A. muciniphila (and consortia including A. muciniphila) is provided in Table 1.

2.2. Impact on Type 2 Diabetes and Glycemic Control

Type 2 diabetes mellitus (T2DM) is another major condition where A. muciniphila shows significant promise. Given the close relationship between obesity and T2DM, many of the metabolic improvements described with A. muciniphila supplementation translate into better glycemic control and insulin function. In high-fat-diet mouse models that develop insulin resistance, A. muciniphila administration improved glucose homeostasis. Treated mice consistently show lower fasting blood glucose levels and enhanced glucose tolerance tests compared to untreated diabetic mice [13,29]. Insulin resistance, a hallmark of T2DM, is alleviated: A. muciniphila-fed mice have higher insulin sensitivity and require less insulin to manage blood sugar [25]. One mechanism for this is a reduction in metabolic endotoxemia—the leakage of LPS from the gut—which otherwise drives inflammation and insulin resistance. By sealing a leaky gut, A. muciniphila prevents LPS-induced inflammation that interferes with insulin signaling [30]. Animal studies also report that A. muciniphila increases the expression of genes involved in energy expenditure and fat oxidation (such as Ucp1 in brown adipose tissue and lipid metabolism genes in the liver), changes that can improve overall insulin sensitivity [31,32,33]. Additionally, A. muciniphila’s production of SCFAs like propionate may stimulate gut hormones that benefit glucose regulation. Propionate released by A. muciniphila has been shown to trigger the secretion of glucagon-like peptide 1 (GLP-1), an incretin hormone that boosts insulin secretion and lowers blood glucose [5]. Experiments indicate that A. muciniphila can augment GLP-1 levels via multiple pathways—not only through SCFAs, but also through a specific protein it produces (called P9) that interacts with host receptors to induce GLP-1 from intestinal L-cells [33]. This dual action on GLP-1 and possibly other gut peptides contributes to better glycemic control in A. muciniphila-treated animals.
The positive impact on diabetic parameters has been borne out in emerging human studies as well. The proof-of-concept trial in 40 overweight/obese individuals mentioned earlier also measured diabetes-related outcomes, since many participants were insulin-resistant [18]. After 3 months of A. muciniphila supplementation, those receiving pasteurized A. muciniphila showed a marked improvement in insulin sensitivity (+28.62% ± 7.02; p = 0.002) and a significant reduction in fasting plasma insulin (−34.08% ± 7.12; p = 0.006) relative to placebo; total cholesterol also fell (−8.68% ± 2.38; p = 0.02). Live A. muciniphila showed directionally favorable but non-significant changes: insulin sensitivity +42.42% ± 20.44 (ns), insulinemia −12.02% ± 4.12 (ns), and total cholesterol −6.70% ± 2.47 (p = 0.088). Fasting glucose did not change dramatically (likely because participants were not frank diabetics), but the lowered insulinemia with pasteurized product points to reduced insulin resistance. These changes imply that the body was able to use insulin more effectively after supplementation (rather than “colonization,” since the pasteurized product is non-viable). The study also reported modest, trend-level effects on anthropometrics: with pasteurized bacteria, body weight −2.27 ± 0.92 kg (p = 0.091), fat mass −1.37 ± 0.82 kg (p = 0.092), hip circumference −2.63 ± 1.14 cm (p = 0.091); with live bacteria, body weight −0.81 ± 0.64 kg, fat mass −0.79 ± 0.47 kg, hip circumference −1.05 ± 0.88 cm (all ns). Participants were instructed to maintain their usual diet and activity, so these metabolic improvements occurred without prescribed lifestyle changes. Overall, the data support a direct metabolic benefit of pasteurized A. muciniphila in this population, with smaller, non-significant shifts in the live arm.
A more recent randomized, double-blind, placebo-controlled phase 2 trial (NCT04797442) in drug-naïve adults with overweight/obese T2DM (n = 58) tested live A. muciniphila (AKK-WST01) once daily for 12 weeks [26]. Overall, weight and HbA1c fell similarly in both arms; however, pre-specified subgroup analyses revealed a clear baseline-dependence: participants with low baseline fecal A. muciniphila exhibited successful strain colonization and significant within-group reductions in body weight, fat mass (including visceral fat), and HbA1c, whereas those with high baseline levels showed little colonization and no clinical benefit. The product was well tolerated. Parallel fecal-transplant experiments in germ-free mice recapitulated the “low-baseline-benefits” pattern, strengthening the biological plausibility of a precision, microbiome-guided use case.
Beyond pure A. muciniphila supplementation, a multicenter, double-blind, placebo-controlled trial (NCT03893422) tested a five-strain consortium (WBF-011) in individuals with T2DM on standard treatment (diet, exercise, and metformin) [28]. The trial had three arms—placebo, WBF-010 (a 3-strain butyrate-focused backbone: Bifidobacterium infantis [Bifidobacterium longum biotype infantis], Clostridium butyricum, Clostridium beijerinckii plus inulin), and WBF-011 (the WBF-010 backbone plus Akkermansia muciniphila and Anaerobutyricum hallii [formerly Eubacterium hallii])—and randomized adults with T2D to capsules twice daily for 12 weeks (n = 76 ITT: placebo 26; WBF-010 27; WBF-011 23; per-protocol n = 58:16/21/21). WBF-011 significantly improved postprandial glucose control versus placebo and produced an HbA1c difference of −0.6% vs. placebo (p = 0.054); within-group, median total postprandial glucose fell 32.5% with WBF-011 (placebo +3.2%). The product was well-tolerated with no major safety signals. Because only the arm that added A. muciniphila and A. hallii to the base consortium improved glycemic endpoints, the authors infer one or both may be necessary, though the design cannot isolate each strain’s contribution. The fact that metformin (a first-line diabetes drug) is known to increase A. muciniphila levels in the gut lends further plausibility to the idea that A. muciniphila contributes to improved glycemic control. In essence, the probiotic cocktail’s success highlights A. muciniphila’s role: by reintroducing this bacterium (and its SCFA-producing partners) into the gut ecosystem, patients achieved better glucose control than with standard care alone.
Preclinical research has also uncovered other diabetes-related benefits of A. muciniphila. For instance, A. muciniphila treatment in mice has been linked to preservation of pancreatic islet cell function [10]. Some studies indicate that maintaining A. muciniphila levels prevents the deterioration of insulin secretion capacity that occurs in diabetes [34]. Moreover, A. muciniphila can influence the metabolism of bile acids and other molecules that affect insulin sensitivity [10]. In murine models, A. muciniphila administration activated the insulin signaling pathway in the liver (PI3K-Akt pathway), which is often impaired in insulin-resistant states [35]. By restoring this pathway’s activity, A. muciniphila helps the liver respond to insulin and regulate glucose production better. A. muciniphila has also been observed to increase levels of endocannabinoids in the gut, compounds that can improve gut barrier function and glucose homeostasis [13]. All these multifaceted actions contribute to an overall antidiabetic effect.
In summary, both animal and human studies strongly support the idea that A. muciniphila supplementation can benefit glycemic control and type 2 diabetes management. It appears to work by improving insulin sensitivity, enhancing gut-derived hormones (e.g., GLP-1), reducing inflammation, and potentially preserving insulin-secreting cell function. Human evidence now includes: (i) a placebo-controlled trial in insulin-resistant volunteers showing metabolic improvements—most notably with pasteurized A. muciniphila—and good tolerability; and (ii) a phase 2 RCT in drug-naïve T2DM demonstrating that the efficacy of live A. muciniphila depends on low baseline Akkermansia abundance (with successful colonization and improvements in weight/adiposity and HbA1c confined to that subgroup). Finally, a multicenter trial of a five-strain consortia that added A. muciniphila to a butyrate-focused backbone improved post-meal glucose and trended to lower HbA1c versus placebo. Collectively, these data suggest A. muciniphila is an emerging adjunct for metabolic disease—especially when guided by baseline microbiome profiling—while larger, longer trials will be needed to define indications, formulation (live vs. pasteurized), and patient selection.

2.3. Impact on Gut Health and Intestinal Function

Given that A. muciniphila naturally dwells in the gut’s mucus layer, its supplementation may have pronounced effects on gut health and integrity. One of the hallmark benefits observed is an improvement in intestinal barrier function. The intestinal barrier is formed by epithelial cells lining the gut, sealed by tight junction proteins, and covered by mucus; it regulates the passage of substances from the gut into circulation. In conditions like obesity, diabetes, or under stress, this barrier often becomes “leaky” (known as increased intestinal permeability), allowing endotoxins and antigens to cross and trigger inflammation [36]. A. muciniphila has demonstrated a remarkable ability to fortify this barrier. In vitro studies showed that A. muciniphila can adhere to intestinal epithelial cells and enhance the integrity of the cell monolayer [17]. This suggests that A. muciniphila may directly interact with the gut lining to stimulate mucus production and tighten junctions between cells. Indeed, rodent studies confirm that A. muciniphila supplementation increases the number of goblet cells (the mucus-producing cells in the gut lining) and the thickness of the mucus layer [4,37]. In obese mice, treatment with A. muciniphila elevated the expression of tight junction proteins like occludin and Zonula occludens-1 in the colon [22,23]. These changes translate to a less permeable intestinal wall—often evidenced by lower levels of circulating LPS in A. muciniphila-treated animals, indicating reduced leakage of endotoxins from the gut. By essentially “patching up” a leaky gut, A. muciniphila creates a healthier intestinal environment and prevents harmful inflammatory triggers from reaching the bloodstream.
A. muciniphila’s impact on gut health also extends to conditions of gut inflammation and disease. Chronic disorders such as inflammatory bowel disease are characterized by disrupted gut barriers and dysregulated immune responses in the intestine. Experimental models of colitis have provided insight into A. muciniphila’s potential benefits and caveats in such contexts. In one study using dextran sulfate sodium (DSS) to induce colitis in mice (a model for ulcerative colitis), administration of A. muciniphila or its purified components significantly attenuated the severity of colitis [38]. Mice receiving A. muciniphila had reduced weight loss, less intestinal bleeding, and improved histological appearance of the colon compared to controls. The beneficial effect was associated with a decrease in pro-inflammatory immune cells infiltrating the colon: treated mice showed fewer macrophages and cytotoxic T cells in colitic lesions, as well as lower levels of inflammatory cytokines like tumor necrosis factor-alpha (TNF-α) and interleukin-1β [39,40]. Additionally, an A. muciniphila-derived outer membrane protein (Amuc_1100) was able to mimic these benefits, suggesting that specific molecules from A. muciniphila can directly interact with the host to quell inflammation [39]. Another protein secreted by A. muciniphila, identified as Amuc_2109 (an enzyme with N-acetylhexosaminidase activity), also protected mice from DSS-induced colitis by upregulating tight junction proteins and downregulating the gut’s inflammatory sensor NLRP3 inflammasome [41]. These findings indicate that A. muciniphila strengthens gut defenses and moderates immune responses during acute intestinal inflammation, at least in some experimental settings.
However, the role of A. muciniphila in gut inflammatory conditions is complex. While the DSS model suggests benefits, another preclinical model of colitis has raised caution. In IL-10 knockout mice, which spontaneously develop IBD-like colitis due to immune dysregulation, supplementation with A. muciniphila was found to exacerbate colonic inflammation in some cases [42]. The absence of the anti-inflammatory cytokine IL-10 creates a highly susceptible environment where the mucus layer is compromised. In such a context, the mucin-degrading activity of A. muciniphila might further erode the already-thin mucus, potentially worsening barrier damage. The study noted that A. muciniphila administration led to earlier onset of colitis in IL-10-deficient mice, suggesting that when the gut barrier is severely impaired, adding A. muciniphila could be detrimental. This highlights an important nuance: A. muciniphila thrives in the mucus layer and usually induces the host to produce more mucus, but if the mucus layer is pathologically thin, its presence might not be beneficial until stability is restored. Conditions like active IBD, severe gut infections, or immediate post-antibiotic gut may not benefit from A. muciniphila in the short term. For example, increasing A. muciniphila during an acute Salmonella infection in mice did not aid recovery and, in fact, correlated with worse inflammation [43]. These results underline that context matters; A. muciniphila is highly beneficial for gut health when it can reinforce a moderately disturbed barrier, but in cases of extreme barrier failure, it might need to be used cautiously. Context-dependent effects of A. muciniphila across preclinical gut models (beneficial and potentially adverse) are synthesized in Table 2.
In terms of general gut homeostasis, A. muciniphila tends to have a normalizing effect on the microbiome and gut environment. Its ability to degrade mucin and produce SCFAs can help support a diverse microbial community. Studies have shown that A. muciniphila can stimulate the growth of other beneficial bacteria by releasing nutrients from mucin metabolism [44]. For instance, the acetate and propionate produced by A. muciniphila serve as fuel for butyrate-producing bacteria, which are crucial for colon health [45]. In supplementation experiments, A. muciniphila has been observed to increase levels of commensal microbes and overall microbial diversity, especially when the gut ecosystem is out of balance [38,40,46,47]. In the human trial, daily A. muciniphila (pasteurized or live) did not drastically alter the overall gut microbiome composition, which is a positive sign that it does not disrupt the native community [18]. Instead, it can coexist and perhaps nudge the ecosystem toward a healthier state by occupying its niche in the mucus layer and contributing beneficial metabolites. Participants in the trial did not experience dysbiosis or opportunistic infections; rather, gut health markers like decreased plasma LPS indicated an improvement.
Beyond pathology, even for general gut wellness, A. muciniphila might confer advantages. Some preliminary evidence in humans links higher A. muciniphila levels to better gut function and bowel regularity. Although formal supplementation studies for conditions like irritable bowel syndrome (IBS) are lacking, one can speculate that A. muciniphila’s barrier-enhancing, anti-inflammatory actions could alleviate certain GI symptoms related to mild gut permeability or immune activation. In summary, A. muciniphila supplementation usually promotes a healthier gut environment: it thickens the protective mucus, tightens cell junctions, reduces pro-inflammatory signaling, and harmonizes interactions between the microbiota and the immune system. These effects protect the gut from insults and may speed recovery from metabolic or dietary stresses. Nonetheless, in extreme cases of gut barrier damage or active colitis, its mucin-degrading nature necessitates careful consideration (a point further discussed later). Overall, the net impact on gut health is positive, positioning A. muciniphila as a guardian of the gut lining and a modulator of gut microbiome homeostasis.

2.4. Impact on Inflammation and Immune Modulation

Chronic inflammation is a common thread linking obesity, diabetes, and gut disorders, and A. muciniphila supplementation has been shown to exert notable anti-inflammatory effects in these contexts. In obese individuals and animals, excess adipose tissue and a leaky gut lead to systemic low-grade inflammation (elevated cytokines like TNF-α, IL-6, etc.), which exacerbates insulin resistance and cardiovascular risk [48]. By sealing the gut barrier, A. muciniphila reduces the influx of inflammatory LPS from the gut, thereby lowering the trigger for systemic inflammation [13,22]. For example, A. muciniphila-treated mice on a high-fat diet had significantly lower plasma LPS levels and reduced expression of inflammatory genes in adipose tissue than untreated mice [13]. These mice also showed a reduction in macrophage infiltration into fat depots, indicating that A. muciniphila curtailed the inflammatory response to obesity. Adipose tissue of treated mice had higher levels of Foxp3+ regulatory T cells (Tregs), which are immune cells that temper inflammation [29]. The increase in Tregs suggests that A. muciniphila actively promotes an anti-inflammatory immune environment. One study found that A. muciniphila induced Tregs in obese mice, correlating with improved glucose tolerance and less adipose inflammation. This immunomodulatory effect extends beyond the gut, as Tregs can circulate and act on multiple tissues to reduce inflammatory signaling.
In systemic metabolic inflammation, A. muciniphila’s impact is also evidenced by changes in liver and blood inflammatory markers. In the mouse model of atherosclerosis (Apoe^−/− mice on a Western diet), A. muciniphila supplementation prevented the diet-induced rise in pro-inflammatory cytokines both in circulation and within atherosclerotic plaques [22]. Treated mice had less macrophage accumulation in their arterial lesions and lower expression of chemokines that recruit immune cells. These anti-inflammatory outcomes were linked with the observed reduction in metabolic endotoxemia and better gut barrier integrity. As the authors concluded, A. muciniphila attenuated atherosclerotic lesions by ameliorating inflammation triggered by metabolic endotoxemia, essentially through gut barrier restoration. This is a powerful demonstration that a probiotic can modulate systemic inflammation enough to impact disease processes in distant organs like blood vessels. Similarly, in the MASLD study in mice, those given A. muciniphila had reduced levels of inflammatory cytokines in the liver and a decreased proportion of pro-inflammatory Th17 cells, alongside an increase in anti-inflammatory Tregs [49]. By shifting the balance from pro-inflammatory Th17 cells to Tregs, A. muciniphila created an environment favoring resolution of inflammation in the liver. These immunological shifts are crucial, as chronic liver inflammation drives the progression from fatty liver to steatohepatitis and fibrosis. Thus, A. muciniphila’s anti-inflammatory effect can halt or slow disease progression.
At the mechanistic level, A. muciniphila appears to interact with the host immune system both locally in the gut and systemically. It has molecular components that engage with pattern recognition receptors on immune cells. One well-studied example is the outer membrane protein Amuc_1100, which is recognized by Toll-like receptor 2 (TLR2) on immune cells [25]. Instead of provoking a strong inflammatory response (as many bacterial proteins might), Amuc_1100 signaling through TLR2 has been shown to have immunomodulatory effects that strengthen the mucus barrier and induce anti-inflammatory cytokines like IL-10 [50]. This helps explain why even pasteurized (non-living) A. muciniphila can reduce inflammation: components (postbiotics) like Amuc_1100 on the bacterial surface remain intact after pasteurization and can still interact with host receptors to drive beneficial immune outcomes. Additionally, A. muciniphila secretes small proteins such as P9 that can cross-talk with immune cells. As noted earlier, P9 was found to bind to ICAM-2 on intestinal L-cells, stimulating GLP-1 release [33]. Intriguingly, ICAM-2 is also expressed on some immune cells, and its engagement can influence immune cell trafficking and activation [51]. While research is ongoing, it is possible that A. muciniphila releases factors that encourage an immune tolerance profile—for example, pushing dendritic cells to favor Treg induction over inflammatory T-cell responses. The observation that A. muciniphila increases IL-10 (an anti-inflammatory cytokine) and Tregs in multiple studies is a consistent theme [29]. See Table 2 for a model-by-model overview of inflammatory contexts in which A. muciniphila has been tested.
Beyond chronic metabolic inflammation, A. muciniphila’s role in immune modulation has implications for other inflammatory or autoimmune conditions. There is emerging interest in how the presence or absence of A. muciniphila might affect disorders like allergies, autoimmune diseases, or even neurological conditions that have an immune component. For instance, some studies have noted that patients with multiple sclerosis or Parkinson’s disease have an unusually high abundance of A. muciniphila in their gut [52]. The significance of this is not fully understood—it could be a compensatory response or a contributor to disease. In animal models of multiple sclerosis (experimental autoimmune encephalomyelitis), manipulating the gut microbiota can alter disease severity, and researchers are investigating if A. muciniphila plays a part in either exacerbating or alleviating such neuroinflammation [53,54,55,56]. It should be cautioned that in conditions like Parkinson’s, where A. muciniphila is already elevated, adding more might not be advisable until we know more [14]. Conversely, in conditions like atopic dermatitis or asthma (which are linked to gut microbiota composition and immune regulation), boosting A. muciniphila might prove beneficial by promoting regulatory immune responses, though direct evidence is still forthcoming.
In summary, the results show that A. muciniphila supplementation generally leads to a reduction in inflammatory markers and a rebalancing of immune responses towards an anti-inflammatory state in metabolic and gut-related diseases. By reducing gut-derived inflammatory triggers and actively engaging immune regulatory pathways, A. muciniphila helps to quell the smoldering inflammation that underpins many chronic conditions. This anti-inflammatory prowess not only improves metabolic indices but could have broader therapeutic implications. However, it is also clear that A. muciniphila is not a one-size-fits-all immunotherapy; its effects can vary depending on the existing immune environment. Most studies paint A. muciniphila as inflammation-attenuating, but a few indicate it could exacerbate inflammation if the gut barrier is severely damaged or in certain dysregulated immune states. These nuances highlight the need for careful application in clinical settings, which we will explore further in the discussion. Nonetheless, the balance of evidence positions A. muciniphila as a valuable ally in reducing chronic inflammation and promoting immune homeostasis.

2.5. Impact on Liver and Cardiovascular Health

Metabolic disorders often harm the liver and the cardiovascular system (promoting atherosclerosis and heart disease) [57]. Encouragingly, A. muciniphila supplementation shows protective effects in these domains, largely through metabolic and anti-inflammatory actions. Beyond weight effects, other metabolic syndrome components also improve with A. muciniphila. Supplemented mice show reduced hepatic steatosis (fatty liver) and improved liver metabolic function [58]. In a mouse model of obesity-related metabolic dysfunction-associated steatotic liver disease (MASLD), treatment with A. muciniphila significantly lowered liver fat and prevented progression of liver injury [49]. Treated mice had lower hepatic inflammation markers, activation of beneficial hepatic regulators (e.g., farnesoid X receptor, FXR), and upregulation of intestinal tight-junction proteins, corresponding with better insulin sensitivity and less weight gain. Together, these findings indicate that A. muciniphila’s anti-obesity actions extend to liver health, potentially weakening the link between obesity and MASLD-related complications.
In the liver specifically, A. muciniphila benefits MASLD both indirectly—by improving obesity and insulin resistance—and directly, via liver-pathway effects. In a high-fat-diet (HFD) mouse model with antibiotic pretreatment, A. muciniphila reduced liver weight/liver index and attenuated hepatic steatosis versus HFD controls, despite identical diets [49]. Liver sections showed fewer/smaller lipid droplets and lower hepatic TG/TC. Systemically, treatment lowered TNF-α, IL-6, IL-17A and raised IL-10, with a shift toward Tregs over Th17. Mechanistically, A. muciniphila increased hepatic FXR while suppressing intestinal FXR and upregulated colonic tight-junction proteins (ZO-1, occludin), consistent with gut-liver axis modulation and barrier reinforcement. Notably, intestinal FGF15 decreased (non-significantly), and the study did not assess MASH/fibrosis endpoints, so anti-fibrotic claims from this experiment are premature. Overall, these data support steatosis improvement and systemic anti-inflammatory effects, with FXR changes differing by tissue.
Across several mouse models, A. muciniphila supplementation has been associated with lower hepatic fibrosis markers. For example, in an HFD + CCl4 injury model, live or pasteurized A. muciniphila (and its extracellular vesicles) reduced collagen deposition and profibrotic genes (e.g., Col1a1, α-SMA, TGF-β) and improved liver histology [59]; in a murine cirrhosis model, administering a commensal A. muciniphila strain improved liver fibrosis (quantified on collagen stains) and hyperammonemia [60]. However, effects are model- and preparation-dependent: in a chronic, diet-induced MASH model (Ldlr−/−. Leiden; 28 weeks), heat-inactivated A. muciniphila did not reduce hepatic collagen/fibrosis despite gut benefits [19]. This suggests anti-fibrotic signals seen in some studies may not generalize across all disease contexts or formulations. While human data on liver outcomes are not yet available, the pilot trial did note reduced blood markers of liver dysfunction in the A. muciniphila-treated participants [18]. This suggests that even over just three months, A. muciniphila might alleviate liver stress or inflammation in overweight individuals. Taken together, these findings point to A. muciniphila as a potential strategy to combat MASLD. By improving metabolic parameters and exerting local effects in the liver (through bile acid metabolism and inflammation reduction), A. muciniphila could halt the early stages of fatty liver disease or even facilitate regression of hepatic fat.
Regarding cardiovascular health, one of the key risk factors is atherosclerosis—the buildup of plaques in arteries, which can lead to heart attacks or strokes. Chronic inflammation and dyslipidemia drive atherosclerosis, and as we have detailed, A. muciniphila favorably influences both factors. In a dedicated Apoe−/− atherosclerosis mouse model, daily oral A. muciniphila for 8 weeks significantly reduced aortic atherosclerotic lesions (area ↓31%, size ↓48%) despite unchanged hypercholesterolemia, compared to control mice on the same high-fat Western diet [22]. Protection tracked with less macrophage infiltration and lower aortic expression of MCP-1, ICAM-1, and TNF-α, alongside improved gut barrier function (↑ ZO-1/occludin, ↓ FITC-dextran permeability) and reduced circulating LPS. Importantly, long-term LPS infusion reversed these benefits, indicating that Akkermansia acts by limiting metabolic endotoxemia and vascular inflammation. Heat-killed bacteria were ineffective in this model. In essence, A. muciniphila maintained the gut barrier, which kept the inflammatory cascade in check, thereby guarding the arteries from the usual damage of a fatty diet. This is a remarkable insight, as it links gut health directly to heart health. It suggests that therapies aimed at the gut microbiome (like A. muciniphila supplements) could complement traditional cardiovascular risk reduction strategies (like statins or diet changes) by targeting the inflammatory aspect of heart disease. Additionally, A. muciniphila’s slight cholesterol-lowering effect observed in humans and its known impact on lipid metabolism genes in mice could have direct cardiovascular benefits [5,18]. Lower total cholesterol and improved lipoprotein profiles reduce the substrate for plaque formation. A concise overview of cardiovascular and hepatic preclinical models is presented in Table 2.
Outside of atherosclerosis, cardiovascular health encompasses blood pressure and heart function, though these have not been deeply studied yet with respect to A. muciniphila. It is known that metabolic syndrome and gut dysbiosis can contribute to hypertension. By improving insulin sensitivity and reducing inflammation, A. muciniphila might indirectly help normalize blood pressure (for example, improved endothelial function due to less inflammation). Human blood pressure data are mixed and largely observational, with small or inconsistent associations so far [61].
Taken together, A. muciniphila supplementation shows significant potential for liver and cardiovascular health. In mice, it mitigates fatty liver/steatosis and attenuates atherosclerosis—the latter via gut-barrier reinforcement and reduced endotoxemia-driven inflammation—even without lowering cholesterol (Apoe−/− model). In humans, a proof-of-concept trial showed improved insulin sensitivity, lower insulinemia, and a modest reduction in total cholesterol with pasteurized A. muciniphila over 3 months, supporting cardiometabolic risk modification but not yet clinical CVD outcomes. For liver disease, multiple mouse models report benefits from steatosis reduction and—in some contexts—lower fibrosis markers, although effects are model- and preparation-dependent (e.g., fibrosis improvement in a cirrhosis model vs. no fibrosis prevention in long-term diet-induced MASH with heat-inactivated cells). If these findings translate into larger trials, A. muciniphila could complement standard strategies by targeting gut-derived inflammation and metabolic dysregulation at their source.

3. Discussion

3.1. Mechanisms of Action

The diverse health benefits of A. muciniphila supplementation observed across obesity, diabetes, gut health, and inflammation can be traced back to several key mechanisms of action. Foremost among these is the strengthening of the gut barrier. A. muciniphila actively reinforces the intestinal lining by increasing mucus production and tight junction integrity. Its presence stimulates goblet cells to secrete more mucin, thickening the protective mucus layer that separates microbes from the intestinal epithelium. This is somewhat paradoxical at first glance—one might expect a mucin-degrading bacterium to thin the mucus layer, but A. muciniphila seems to trigger a compensatory response from the host to produce even more mucus, resulting in an overall enhancement of the barrier. Additionally, A. muciniphila or its components can upregulate tight junction proteins between epithelial cells, as shown in mice and cell culture models. The net effect is reduced intestinal permeability. A less “leaky” gut means fewer endotoxins like LPS escaping into circulation. Since metabolic endotoxemia is a driver of systemic inflammation and insulin resistance, A. muciniphila’s ability to reduce LPS translocation is fundamental to its benefits. By resealing the gut barrier, A. muciniphila cuts off the trigger for the cascade of inflammation that links an unhealthy gut to obesity-related disorders. This gut barrier fortification is arguably the cornerstone of A. muciniphila’s action—it creates a healthier separation between the gut microbiota and the host, thereby maintaining immune equilibrium and metabolic homeostasis.
Another mechanism is the production of bioactive metabolites. A. muciniphila feeds on mucin and, in the process, releases SCFAs (like acetate and propionate) and possibly other metabolites such as amino acids and indole derivatives. SCFAs have multiple beneficial effects on the host: they serve as energy sources for colon cells, lower gut pH to inhibit pathogens, and act as signaling molecules [62]. Propionate and acetate produced by A. muciniphila can bind to free fatty acid receptors (such as FFAR2/3) on enteroendocrine cells, stimulating the secretion of satiety and metabolic hormones [33,63,64]. In particular, propionate is known to induce GLP-1 and peptide YY (PYY) release, hormones that improve insulin secretion, reduce appetite, and slow gut motility (contributing to weight management). The elevation of GLP-1 observed with A. muciniphila supplementation in mice aligns with this mechanism, as do the improved glucose tolerance and reduced food intake in some studies. Moreover, A. muciniphila’s metabolic byproducts may influence gene expression in tissues. For example, SCFAs can act as histone deacetylase inhibitors, modulating gene transcription in the liver and adipose tissue to favor fat burning over storage [65]. The metabolite-driven activation of the AMPK pathway in the liver (noted in one mouse study) suggests improved energy sensing and reduction in fat synthesis [32]. Evidence that A. muciniphila raises circulating phospholipids or lowers plasma branched-chain amino acids (BCAAs) in humans is indirect; its bacterial phospholipid can signal to the host, and BCAAs are linked to insulin resistance, but direct Akkermansia-driven changes in these plasma metabolites remain to be demonstrated [66]. Thus, via its metabolic outputs, A. muciniphila communicates with the host’s physiology, essentially extending the functionality of our own metabolism.
Direct host–microbe signaling molecules from A. muciniphila represent another mechanism. We have mentioned proteins like Amuc_1100 (an outer membrane protein) and P9 (a small secreted protein), which have specific interactions with host receptors. Amuc_1100, identified by Plovier et al., can bind to TLR2 on intestinal epithelial and immune cells, triggering pathways that enhance mucus production and anti-inflammatory IL-10 release, without causing the pro-inflammatory effects that typical TLR2 agonists (like certain peptidoglycans) might [25]. This selective activation helps fine-tune the immune system toward tolerance. P9, as discovered by Yoon et al., binds to ICAM-2 on L cells, driving GLP-1 secretion and thus improving glucose handling [33]. These are examples of A. muciniphila essentially delivering therapeutic molecules to the host. Importantly, pasteurization (mild heat-killing) of A. muciniphila preserves these proteins, which explains why pasteurized A. muciniphila was effective in the human trial [18]. The bacterial cells need not be alive to confer benefits; they can act as a rich package of beneficial molecules—a concept known as postbiotics. In fact, the successful use of pasteurized A. muciniphila in humans led to its designation as a novel food ingredient (since it is not a traditional probiotic, being non-living) [14]. This could mark a paradigm shift: leveraging bacterial components (like surface proteins, membrane fragments, metabolites) to achieve health outcomes, which can simplify regulatory approval and safety profiles (no risk of live infection).

3.2. Clinical Studies and Translational Insights

Translation of A. muciniphila from bench to bedside is in early stages, but initial trials are informative. The 2019 Belgian randomized trial established that daily 1010 cells (live or pasteurized) is feasible and well tolerated, with no bacterium-attributed adverse events, and that pasteurized cells outperformed placebo on several metabolic endpoints (e.g., insulin sensitivity, insulinemia, total cholesterol) in overweight/obese insulin-resistant adults [18]. Because A. muciniphila is a commensal of the human gut, this amounts to boosting a native species rather than introducing an exogenous microbe. Regulatory momentum followed: in 2021, the European Food Safety Authority (EFSA) concluded pasteurized A. muciniphila is safe for adult consumption up to ~3.4 × 1010 cells/day, provided viable cells remain < 10 CFU/g, leading to EU market authorization—facilitating development of heat-inactivated, lyophilized formulations that are easier to handle than live anaerobes [14]. In 2025, EFSA extended safety to adolescents with age-specific dose caps; safety in pregnancy/lactation remains unestablished. Separately, a US multicenter RCT in type 2 diabetes tested a five-strain consortium including A. muciniphila (WBF-011), dosed twice daily for 12 weeks, which improved postprandial glucose AUC and reduced A1c versus placebo with no safety signals, supporting the concept that A. muciniphila can synergize with other anaerobes in disease-focused probiotics [28]. Finally, diet remains a key lever: while human trials to date typically do not restrict fiber, animal work shows prebiotic fibers (e.g., fructooligosaccharides/inulin) can markedly increase A. muciniphila and improve metabolic outcomes, motivating future synbiotic strategies that pair A. muciniphila with targeted prebiotics [13]. A consolidated snapshot of human trials—including design, dose/formulation, and key readouts—appears in Table 1.
Taken together, current human trials are small and short (about 3 months) and use heterogeneous formulations—single-strain live, single-strain pasteurized, and multi-strain consortia—limiting cross-study comparability and the strength of inferences about efficacy. The strongest signal to date comes from a single-center RCT in insulin-resistant adults where pasteurized cells improved insulin sensitivity and lipids, whereas weight changes were modest (3 months) [18]. By contrast, a Phase 2 RCT of a live strain in overweight/obese T2D found no overall between-group effect on weight or HbA1c, but benefit in the pre-specified subgroup with low baseline Akkermansia—highlighting baseline-dependent responses and the need for stratified designs [26]. Multi-strain data (e.g., WBF-011) suggest glycemic improvements in T2D, yet attribution to Akkermansia specifically is not possible and concomitant standard care/dietary guidance can confound outcomes, underscoring the need for larger, longer RCTs with harmonized endpoints and pre-planned subgroup analyses. Because obesity, T2D, and metabolic syndrome span multiple phenotypes and severities, future trials should prospectively stratify by these features to reduce bias and clarify who benefits.

3.3. Commercialization and Available Supplements

Commercial products featuring Akkermansia muciniphila fall into two broad categories: (1) live-strain probiotics, and (2) pasteurized (postbiotic) preparations. Consistent with the ISAPP consensus, postbiotics are preparations of inanimate microorganisms and/or their components that confer a health benefit, whereas probiotics contain live microorganisms administered in adequate amounts to confer a benefit [67]. In the United States, Pendulum Therapeutics markets (i) Pendulum Glucose Control (a multi-strain formulation that includes A. muciniphila) and (ii) Pendulum Akkermansia (stand-alone live A. muciniphila). The Glucose Control formulation has been evaluated in a double-blind, placebo-controlled trial in adults with type 2 diabetes, demonstrating safety and improvements in post-prandial glycemia (and A1C in secondary analyses), though sample size was modest and strain-specific effects cannot be fully isolated in a multi-strain product [28]. These products are sold in the U.S. as dietary supplements and are not FDA-approved drugs; claims are therefore limited to structure–function language.
In the European Union, The Akkermansia Company (A-Mansia) commercializes pasteurized A. muciniphila (Akkermansia MucT™) under the EU Novel Food framework. EFSA’s 2021 scientific opinion concluded the ingredient is safe for adults at specified doses in food supplements and foods for special medical purposes (FSMPs) (pregnant/lactating women excluded pending data). In 2025, EFSA issued an extension of use supporting defined maximum daily doses in adolescents (12 to <14 years and 14 to <18 years). Labeling in the EU reflects these conditions of use, including heat-treated (non-viable) status and dose limits.
Live products (e.g., Pendulum) and pasteurized products (e.g., A-Mansia) differ in (i) regulatory pathway (U.S. dietary supplement vs. EU Novel Food authorization), (ii) formulation/handling (cold-chain and viability considerations for live strains vs. heat-stabilized postbiotics), and (iii) evidence base (multi-strain, condition-specific trials vs. ingredient-level safety authorizations and emerging efficacy data). These differences should guide counseling on product selection, expected effects, and dose/formulation adherence.

3.4. Limitations and Considerations

Despite the excitement, there are several considerations and open questions regarding A. muciniphila supplementation. First, individual variability: not everyone may respond the same way to A. muciniphila. Baseline microbiome composition could influence its colonization. If someone already has abundant A. muciniphila, supplementing more might have diminishing returns. Conversely, someone with none (perhaps due to a long-term low-fiber diet or antibiotic use) might benefit greatly, but the bacteria will need a hospitable environment (sufficient mucin or prebiotics) to establish. There is also the question of long-term safety and effects. The trials so far have been short-term (3 months). Long-term supplementation is presumed safe given A. muciniphila’s commensal nature, but we should monitor whether the gut adapts (e.g., does endogenous mucin production continually increase, and is that always good?). Extremely high levels of A. muciniphila might conceivably alter the gut ecosystem in unknown ways. As noted in the “critical perspective” review, excessive enrichment of A. muciniphila might not be beneficial in every circumstance. For example, if someone is on a mucin-free diet or has a genetic mucus disorder, flooding the gut with a mucin-degrader could hypothetically cause problems.
Obesity, type 2 diabetes, and metabolic syndrome are clinically heterogeneous—they vary by adiposity distribution/phenotype (e.g., “metabolically healthy” vs. “unhealthy” obesity), inflammatory tone, insulin secretory/β-cell defects, and cardiometabolic clustering—so response to A. muciniphila may differ across endotypes and severity. Contemporary frameworks emphasize a cardiovascular–kidney–metabolic (CKM) spectrum rather than siloed diseases, underscoring diverse trajectories and competing risks in the same patient. Accordingly, trials should pre-specify phenotype/phase (e.g., insulin-resistant but non-diabetic vs. drug-naïve T2D), stratify by baseline metabolic features (waist/visceral adiposity, lipid pattern, glycemia/A1c, blood pressure) and co-morbid CKD/CVD, and report outcomes by subgroup (including metabolically healthy vs. unhealthy obesity), to avoid averaging away clinically meaningful effects. This heterogeneity—recognized in ADA Standards, AHA CKM advisories, and the obesity/MASLD phenotype literature—should inform both study design and clinical interpretation of A. muciniphila interventions [68].
Second, formulation heterogeneity (live vs. pasteurized; single-strain vs. consortia) and host context are likely effect modifiers. Signals for pasteurized preparations on insulin sensitivity and lipids have not yet been mirrored consistently with live strains across populations and endpoints [18]. Moreover, baseline Akkermansia abundance appears consequential: a Phase 2 RCT reported that individuals starting low benefited (weight, fat mass, HbA1c), whereas those already high did not, suggesting a precision-use paradigm (e.g., baseline profiling or enrichment strategies) [26]. Third, concomitant therapies and diets introduce confounding—for example, metformin is repeatedly associated with higher Akkermansia in humans, and RCTs often include lifestyle counseling or standard-of-care adjustments that can mask between-group differences; future trials should pre-specify drug–microbe and diet–microbe interactions and stratify or adjust accordingly [69].
Population exceptions warrant personalized guidance. In IBD, A. muciniphila is often reduced, and some models show barrier support, but disease activity and mucosal integrity likely modulate effects; prudence suggests focusing on remission-maintenance rather than active flares until more human data accrue. In neurologic conditions (e.g., Parkinson’s disease), higher Akkermansia abundance is consistently observed, yet causality is unresolved; supplementation in such groups should be studied in trials before routine use. Finally, with respect to blood pressure, human evidence remains preliminary and heterogeneous—current data are drawn largely from observational cohorts and small interventional paradigms (e.g., dietary programs that alter multiple variables alongside the microbiome), and there are no randomized trials of A. muciniphila monotherapy showing antihypertensive efficacy, so any impact on blood pressure should be considered speculative pending dedicated studies. Overall, current evidence is strongest in metabolically unhealthy but otherwise generally well adults, while special populations should be approached cautiously pending long-term and condition-specific data. Practical implications for study design and patient selection are compiled in Table 3.

3.5. Future Directions

Larger, confirmatory trials of Akkermansia muciniphila in metabolic disease are now underway, including randomized studies in hyperglycemia/overweight and in weight maintenance after diet-induced loss (e.g., NCT05114018, NCT05417360); these trials will enable dose-finding and may compare live vs. pasteurized directly, extending the initial 3-month Belgian proof-of-concept that showed safety and metabolic signals—particularly with pasteurized cells (e.g., NCT05114018) [70]. Key endpoints will expand from surrogate measures to clinical outcomes (e.g., A1c, body weight trajectories). Given that metformin consistently enriches Akkermansia in humans and that GLP-1R agonists are associated with microbiome shifts (often including Akkermansia), it is reasonable to test combination strategies (microbe + drug) and diet–microbe programs prospectively, while recognizing that synergy remains unproven. Beyond obesity/T2D, MASLD represents a high-priority target (animal data promising; human RCTs needed), and oncology trials may examine whether adding A. muciniphila improves immune checkpoint inhibitor responses, given repeated human associations between baseline Akkermansia and PD-1/PD-L1 efficacy. For polycystic ovary syndrome (PCOS), human microbiome studies report dysbiosis involving Akkermansia, but interventional data and IBD-risk linkage remain uncertain, warranting cautious exploration.
Mechanistically, discovery will likely accelerate. Amuc_1100 (outer-membrane, heat-stable; TLR2 signaling) and P9 (a GLP-1–inducing secreted protein binding ICAM-2) exemplify how A. muciniphila communicates with the host. Extracellular vesicles (EVs) and pasteurized whole-cell postbiotics already show robust preclinical or early clinical activity. A key open question is how far cell-free fractions (culture supernatants) can reproduce benefits in mammals/humans—early model systems are encouraging, but translational studies are needed. These lines of work point toward drug-like, defined postbiotics that could be trialed where whole-cell administration is impractical. Planned/ongoing clinical questions and how they map onto existing evidence are cross-referenced in Table 1 (human trials) and Table 3 (practical considerations).

4. Methods

This article is a narrative review aimed at integrating mechanistic, translational, and early clinical evidence on Akkermansia muciniphila with an emphasis on cardiometabolic and gut-barrier outcomes without attempting a systematic review or meta-analysis. A literature search was conducted using PubMed to identify relevant publications on Akkermansia muciniphila supplementation through September 2025. Search terms included “Akkermansia muciniphila”, “supplementation”, “probiotic”, “obesity”, “diabetes”, “inflammation”, “gut barrier”, and related keywords. Priority was given to peer-reviewed studies and meta-analyses focusing on human clinical trials. Animal and in vitro studies were included when they elucidated mechanisms or provided important proof-of-concept data. Key reference lists were also scanned to capture additional relevant studies.
We included preclinical studies (e.g., in animal models) that established mechanisms of action, as well as all available human trials of A. muciniphila or A. muciniphila-containing formulations. Only studies indexed in PubMed and published in English were considered. In total, dozens of articles were reviewed, and those most pertinent to A. muciniphila’s effects on obesity, type 2 diabetes, gut health (including gut inflammation), and related conditions were synthesized for this manuscript. We excluded studies not indexed in PubMed and non-English publications; preprints and other non-peer-reviewed reports. Given the narrative aim, we performed no quantitative pooling, no formal risk-of-bias grading, and no pre-registered protocol. To encourage transparent reporting for narrative syntheses, we aligned with good-practice guidance for narrative reviews (e.g., clearly stated aims, rationale for article selection, and balanced presentation of evidence and limitations) [71].

5. Conclusions

Akkermansia muciniphila has progressed from commensal curiosity to a plausible therapeutic adjunct. In preclinical systems, live and pasteurised preparations—and defined components such as Amuc_1100—consistently strengthen barrier function, modulate inflammation, and improve metabolic readouts. Early human data echo parts of this signal but remain limited in size, duration, and comparability. In a single-center, 3-month RCT in overweight/obese insulin-resistant adults, pasteurised A. muciniphila was safe and well tolerated and improved insulin sensitivity and lowered total cholesterol versus placebo, while anthropometric changes trended but were not statistically significant. A multicenter RCT of a five-strain consortium that included A. muciniphila improved post-prandial glucose control and A1c in type 2 diabetes, but effects cannot be attributed to Akkermansia specifically. A Phase 2 RCT with a live single strain in overweight/obese T2D reported no overall between-group benefit on weight or HbA1c, with metabolic improvements confined to participants with low baseline Akkermansia—a pattern suggesting baseline-dependent response and a need for stratified designs.
Commercial translation is underway, but should be interpreted within regulatory boundaries. In the EU, pasteurised A. muciniphila is authorized as a novel food for adults (finished products <10 CFU/g viable), with a 2025 EFSA extension specifying adolescent use at capped doses; this has enabled room-temperature postbiotic formulations. In the US, a multistrain product containing A. muciniphila is marketed as a dietary supplement and is supported by the above RCT, which nonetheless reflects consortia rather than single-strain effects. Going forward, responsible integration will require larger, longer randomized trials that: (i) compare live vs. pasteurized formulations head-to-head; (ii) pre-specify precision-use hypotheses (e.g., baseline abundance, colonization) and adjust for co-interventions; and (iii) assess hard outcomes (clinically meaningful weight change, HbA1c trajectories, MASLD endpoints). Given that metformin is associated with higher Akkermansia abundance in humans—and that many trials include lifestyle counseling—future studies should account for drug–microbe and diet–microbe interactions to minimize confounding. Inter-individual variability (phenotype, comorbidities) and co-medications are likely effect modifiers, reinforcing the need for baseline profiling and documentation of colonization when interpreting outcomes.
In sum, A. muciniphila—particularly in postbiotic (pasteurized) formats with favorable logistics—appears promising as an adjunct for metabolic health. However, formulation differences, host context, and trial heterogeneity temper certainty at this stage. A measured conclusion is warranted: enthusiasm should be paired with condition-specific trials, careful patient selection, and transparent communication of what is known—and not yet known.

Author Contributions

A.F. and M.H.C.N. contributed equally to conceptualization, methodology, investigation, resources, data curation, writing—original draft preparation, and writing—review and editing; supervision, A.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

Author Minh Hien Chau Nguyen was employed by the company CVS Health. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
A. muciniphilaAkkermansia muciniphila
A1cGlycated hemoglobin (HbA1c). (Used interchangeably with HbA1c in the text.)
AMPKAMP-activated protein kinase
Apoe−/−Apolipoprotein E knockout (mouse model)
ASTAspartate aminotransferase
BCAA(s)Branched-chain amino acid(s)
CCl4Carbon tetrachloride
CFUColony-forming units
CONSORTConsolidated Standards of Reporting Trials
CVDCardiovascular disease
DSSDextran sulfate sodium
EFSAEuropean Food Safety Authority
EUEuropean Union
EV(s)Extracellular vesicle(s)
FFAR2/3Free fatty acid receptor 2/3
FGF15Fibroblast growth factor 15
FXRFarnesoid X receptor
GIGastrointestinal
GLP-1Glucagon-like peptide-1
GLP-1RGLP-1 receptor (appears in “GLP-1R agonists”)
HbA1cHemoglobin A1c (glycated hemoglobin) (Also appears as A1c.)
HFDHigh-fat diet
IBDInflammatory bowel disease
IBSIrritable bowel syndrome
ICAM-2Intercellular adhesion molecule-2
IL-10/IL-6/IL-17AInterleukin-10/-6/-17A
ITTIntent-to-treat (analysis set)
LPSLipopolysaccharide
MASLDMetabolic dysfunction-associated steatotic liver disease
MASHMetabolic dysfunction-associated steatohepatitis
MCPMonocyte chemoattractant protein
NLRP3NOD-like receptor family pyrin domain containing 3 (inflammasome sensor)
PCOSPolycystic ovary syndrome
PI3K-AktPhosphoinositide-3-kinase/AKT signaling pathway
PRISMAPreferred Reporting Items for Systematic Reviews and Meta-Analyses
PYYPeptide YY
RCTRandomized controlled trial
SCFA(s)Short-chain fatty acid(s)
T2D/T2DMType 2 diabetes/Type 2 diabetes mellitus
TCTotal cholesterol
TGTriglycerides
Th17T helper 17 cell
TLR2Toll-like receptor 2
TNFTumor necrosis factor
Treg(s)Regulatory T cell(s)
USUnited States
ZO-1Zonula occludens-1

References

  1. Ioannou, A.; Berkhout, M.D.; Geerlings, S.Y.; Belzer, C. Akkermansia muciniphila: Biology, microbial ecology, host interactions and therapeutic potential. Nat. Rev. Microbiol. 2025, 23, 162–177. [Google Scholar] [CrossRef]
  2. Cani, P.D.; Depommier, C.; Derrien, M.; Everard, A.; de Vos, W.M. Akkermansia muciniphila: Paradigm for next-generation beneficial microorganisms. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 625–637. [Google Scholar] [CrossRef]
  3. Derrien, M.; Vaughan, E.E.; Plugge, C.M.; de Vos, W.M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 2004, 54 Pt 5, 1469–1476. [Google Scholar] [CrossRef]
  4. van der Lugt, B.; van Beek, A.A.; Aalvink, S.; Meijer, B.; Sovran, B.; Vermeij, W.P.; Brandt, R.M.C.; de Vos, W.M.; Savelkoul, H.F.J.; Steegenga, W.T.; et al. Akkermansia muciniphila ameliorates the age-related decline in colonic mucus thickness and attenuates immune activation in accelerated aging Ercc1 (-/Delta7) mice. Immun. Ageing 2019, 16, 6. [Google Scholar] [CrossRef]
  5. Li, J.; Yang, G.; Zhang, Q.; Liu, Z.; Jiang, X.; Xin, Y. Function of Akkermansia muciniphila in type 2 diabetes and related diseases. Front. Microbiol. 2023, 14, 1172400. [Google Scholar] [CrossRef]
  6. Zhou, Q.; Zhang, Y.; Wang, X.; Yang, R.; Zhu, X.; Zhang, Y.; Chen, C.; Yuan, H.; Yang, Z.; Sun, L. Gut bacteria Akkermansia is associated with reduced risk of obesity: Evidence from the American Gut Project. Nutr. Metab. 2020, 17, 90. [Google Scholar] [CrossRef]
  7. Santacruz, A.; Collado, M.C.; Garcia-Valdes, L.; Segura, M.T.; Martin-Lagos, J.A.; Anjos, T.; Marti-Romero, M.; Lopez, R.M.; Florido, J.; Campoy, C.; et al. Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Br. J. Nutr. 2010, 104, 83–92. [Google Scholar] [CrossRef]
  8. Karlsson, C.L.; Onnerfalt, J.; Xu, J.; Molin, G.; Ahrne, S.; Thorngren-Jerneck, K. The microbiota of the gut in preschool children with normal and excessive body weight. Obesity 2012, 20, 2257–2261. [Google Scholar] [CrossRef]
  9. Zhang, X.; Shen, D.; Fang, Z.; Jie, Z.; Qiu, X.; Zhang, C.; Chen, Y.; Ji, L. Human gut microbiota changes reveal the progression of glucose intolerance. PLoS ONE 2013, 8, e71108. [Google Scholar] [CrossRef]
  10. Zhang, J.; Ni, Y.; Qian, L.; Fang, Q.; Zheng, T.; Zhang, M.; Gao, Q.; Zhang, Y.; Ni, J.; Hou, X.; et al. Decreased Abundance of Akkermansia muciniphila Leads to the Impairment of Insulin Secretion and Glucose Homeostasis in Lean Type 2 Diabetes. Adv. Sci. 2021, 8, e2100536. [Google Scholar] [CrossRef]
  11. Li, Q.; Chang, Y.; Zhang, K.; Chen, H.; Tao, S.; Zhang, Z. Implication of the gut microbiome composition of type 2 diabetic patients from northern China. Sci. Rep. 2020, 10, 5450. [Google Scholar] [CrossRef]
  12. Xu, Z.; Jiang, W.; Huang, W.; Lin, Y.; Chan, F.K.L.; Ng, S.C. Gut microbiota in patients with obesity and metabolic disorders—A systematic review. Genes. Nutr. 2022, 17, 2. [Google Scholar] [CrossRef]
  13. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef]
  14. Chiantera, V.; Lagana, A.S.; Basciani, S.; Nordio, M.; Bizzarri, M. A Critical Perspective on the Supplementation of Akkermansia muciniphila: Benefits and Harms. Life 2023, 13, 1247. [Google Scholar] [CrossRef] [PubMed]
  15. Rodrigues, V.F.; Elias-Oliveira, J.; Pereira, I.S.; Pereira, J.A.; Barbosa, S.C.; Machado, M.S.G.; Carlos, D. Akkermansia muciniphila and Gut Immune System: A Good Friendship That Attenuates Inflammatory Bowel Disease, Obesity, and Diabetes. Front. Immunol. 2022, 13, 934695. [Google Scholar] [CrossRef]
  16. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef]
  17. Reunanen, J.; Kainulainen, V.; Huuskonen, L.; Ottman, N.; Belzer, C.; Huhtinen, H.; de Vos, W.M.; Satokari, R. Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity of the Epithelial Cell Layer. Appl. Environ. Microbiol. 2015, 81, 3655–3662. [Google Scholar] [CrossRef]
  18. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef]
  19. Baethge, C.; Goldbeck-Wood, S.; Mertens, S. SANRA-a scale for the quality assessment of narrative review articles. Res. Integr. Peer Rev. 2019, 4, 5. [Google Scholar] [CrossRef]
  20. Morrison, M.C.; Gart, E.; Duyvenvoorde, W.V.; Snabel, J.; Nielsen, M.J.; Leeming, D.J.; Menke, A.; Kleemann, R. Heat-Inactivated Akkermansia muciniphila Improves Gut Permeability but Does Not Prevent Development of Non-Alcoholic Steatohepatitis in Diet-Induced Obese Ldlr−/−.Leiden Mice. Int. J. Mol. Sci. 2022, 23, 2325. [Google Scholar] [CrossRef]
  21. Liu, E.; Ji, X.; Zhou, K. Akkermansia muciniphila for the Prevention of Type 2 Diabetes and Obesity: A Meta-Analysis of Animal Studies. Nutrients 2024, 16, 3440. [Google Scholar] [CrossRef]
  22. Katiraei, S.; de Vries, M.R.; Costain, A.H.; Thiem, K.; Hoving, L.R.; van Diepen, J.A.; Smits, H.H.; Bouter, K.E.; Rensen, P.C.N.; Quax, P.H.A.; et al. Akkermansia muciniphila Exerts Lipid-Lowering and Immunomodulatory Effects without Affecting Neointima Formation in Hyperlipidemic APOE*3-Leiden.CETP Mice. Mol. Nutr. Food Res. 2020, 64, e1900732. [Google Scholar] [CrossRef]
  23. Li, J.; Lin, S.; Vanhoutte, P.M.; Woo, C.W.; Xu, A. Akkermansia Muciniphila Protects Against Atherosclerosis by Preventing Metabolic Endotoxemia-Induced Inflammation in Apoe−/− Mice. Circulation 2016, 133, 2434–2446. [Google Scholar] [CrossRef]
  24. Ashrafian, F.; Shahriary, A.; Behrouzi, A.; Moradi, H.R.; Keshavarz Azizi Raftar, S.; Lari, A.; Hadifar, S.; Yaghoubfar, R.; Ahmadi Badi, S.; Khatami, S.; et al. Akkermansia muciniphila-Derived Extracellular Vesicles as a Mucosal Delivery Vector for Amelioration of Obesity in Mice. Front. Microbiol. 2019, 10, 2155. [Google Scholar] [CrossRef]
  25. Ashrafian, F.; Keshavarz Azizi Raftar, S.; Shahryari, A.; Behrouzi, A.; Yaghoubfar, R.; Lari, A.; Moradi, H.R.; Khatami, S.; Omrani, M.D.; Vaziri, F.; et al. Comparative effects of alive and pasteurized Akkermansia muciniphila on normal diet-fed mice. Sci. Rep. 2021, 11, 17898. [Google Scholar] [CrossRef]
  26. Plovier, H.; Everard, A.; Druart, C.; Depommier, C.; Van Hul, M.; Geurts, L.; Chilloux, J.; Ottman, N.; Duparc, T.; Lichtenstein, L.; et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat. Med. 2017, 23, 107–113. [Google Scholar] [CrossRef]
  27. Zhang, Y.; Liu, R.; Chen, Y.; Cao, Z.; Liu, C.; Bao, R.; Wang, Y.; Huang, S.; Pan, S.; Qin, L.; et al. Akkermansia muciniphila supplementation in patients with overweight/obese type 2 diabetes: Efficacy depends on its baseline levels in the gut. Cell Metab. 2025, 37, 592–605 e596. [Google Scholar] [CrossRef]
  28. Aalipanah, E.; Askarpour, M.; Eskandari, M.H.; Zare, M.; Famouri, M.; Bedeltavana, A.; Mohsenpour, M.A.; Sohrabi, Z. Comparing the effects of yogurt containing Akkermansia muciniphilia postbiotic with yogurt containing Lactobacillus rhamnosus postbiotic on body composition, biochemical indices, appetite, and depression scores in overweight or obese adults: A randomized, double-blind, controlled clinical trial. Clin. Nutr. ESPEN 2025, 68, 438–446. [Google Scholar] [CrossRef]
  29. Shin, N.R.; Lee, J.C.; Lee, H.Y.; Kim, M.S.; Whon, T.W.; Lee, M.S.; Bae, J.W. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014, 63, 727–735. [Google Scholar] [CrossRef]
  30. Cani, P.D.; de Vos, W.M. Next-Generation Beneficial Microbes: The Case of Akkermansia muciniphila. Front. Microbiol. 2017, 8, 1765. [Google Scholar] [CrossRef]
  31. Deng, L.; Ou, Z.; Huang, D.; Li, C.; Lu, Z.; Liu, W.; Wu, F.; Nong, C.; Gao, J.; Peng, Y. Diverse effects of different Akkermansia muciniphila genotypes on Brown adipose tissue inflammation and whitening in a high-fat-diet murine model. Microb. Pathog. 2020, 147, 104353. [Google Scholar] [CrossRef] [PubMed]
  32. Rao, Y.; Kuang, Z.; Li, C.; Guo, S.; Xu, Y.; Zhao, D.; Hu, Y.; Song, B.; Jiang, Z.; Ge, Z.; et al. Gut Akkermansia muciniphila ameliorates metabolic dysfunction-associated fatty liver disease by regulating the metabolism of L-aspartate via gut-liver axis. Gut Microbes 2021, 13, 1927633. [Google Scholar] [CrossRef]
  33. Yoon, H.S.; Cho, C.H.; Yun, M.S.; Jang, S.J.; You, H.J.; Kim, J.H.; Han, D.; Cha, K.H.; Moon, S.H.; Lee, K.; et al. Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nat. Microbiol. 2021, 6, 563–573. [Google Scholar] [CrossRef]
  34. Perraudeau, F.; McMurdie, P.; Bullard, J.; Cheng, A.; Cutcliffe, C.; Deo, A.; Eid, J.; Gines, J.; Iyer, M.; Justice, N.; et al. Improvements to postprandial glucose control in subjects with type 2 diabetes: A multicenter, double blind, randomized placebo-controlled trial of a novel probiotic formulation. BMJ Open Diabetes Res. Care 2020, 8, e001319. [Google Scholar] [CrossRef]
  35. Yan, S.; Chen, L.; Li, N.; Wei, X.; Wang, J.; Dong, W.; Wang, Y.; Shi, J.; Ding, X.; Peng, Y. Effect of Akkermansia muciniphila on pancreatic islet beta-cell function in rats with prediabetes mellitus induced by a high-fat diet. Bioresour. Bioprocess. 2024, 11, 51. [Google Scholar] [CrossRef]
  36. Xia, J.; Lv, L.; Liu, B.; Wang, S.; Zhang, S.; Wu, Z.; Yang, L.; Bian, X.; Wang, Q.; Wang, K.; et al. Akkermansia muciniphila Ameliorates Acetaminophen-Induced Liver Injury by Regulating Gut Microbial Composition and Metabolism. Microbiol. Spectr. 2022, 10, e0159621. [Google Scholar] [CrossRef]
  37. Camilleri, M. Is intestinal permeability increased in obesity? A review including the effects of dietary, pharmacological and surgical interventions on permeability and the microbiome. Diabetes Obes. Metab. 2023, 25, 325–330. [Google Scholar] [CrossRef]
  38. Yu, J.; Liu, T.; Gao, Z.; Liu, R.; Wang, Z.; Chen, Y.; Cao, J.; Dong, Y. Akkermansia muciniphila Colonization Alleviating High Fructose and Restraint Stress-Induced Jejunal Mucosal Barrier Disruption. Nutrients 2022, 14, 3164. [Google Scholar] [CrossRef]
  39. Bian, X.; Wu, W.; Yang, L.; Lv, L.; Wang, Q.; Li, Y.; Ye, J.; Fang, D.; Wu, J.; Jiang, X.; et al. Administration of Akkermansia muciniphila Ameliorates Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice. Front. Microbiol. 2019, 10, 2259. [Google Scholar] [CrossRef]
  40. Wang, L.; Tang, L.; Feng, Y.; Zhao, S.; Han, M.; Zhang, C.; Yuan, G.; Zhu, J.; Cao, S.; Wu, Q.; et al. A purified membrane protein from Akkermansia muciniphila or the pasteurised bacterium blunts colitis associated tumourigenesis by modulation of CD8(+) T cells in mice. Gut 2020, 69, 1988–1997. [Google Scholar] [CrossRef]
  41. Zhang, H.; Pan, Y.; Jiang, Y.; Chen, M.; Ma, X.; Yu, X.; Ren, D.; Jiang, B. Akkermansia muciniphila ONE effectively ameliorates dextran sulfate sodium (DSS)-induced ulcerative colitis in mice. NPJ Sci. Food 2024, 8, 97. [Google Scholar] [CrossRef]
  42. Qian, K.; Chen, S.; Wang, J.; Sheng, K.; Wang, Y.; Zhang, M. A beta-N-acetylhexosaminidase Amuc_2109 from Akkermansia muciniphila protects against dextran sulfate sodium-induced colitis in mice by enhancing intestinal barrier and modulating gut microbiota. Food Funct. 2022, 13, 2216–2227. [Google Scholar] [CrossRef]
  43. Seregin, S.S.; Golovchenko, N.; Schaf, B.; Chen, J.; Pudlo, N.A.; Mitchell, J.; Baxter, N.T.; Zhao, L.; Schloss, P.D.; Martens, E.C.; et al. NLRP6 Protects Il10(−/−) Mice from Colitis by Limiting Colonization of Akkermansia muciniphila. Cell Rep. 2017, 19, 733–745. [Google Scholar] [CrossRef]
  44. Ganesh, B.P.; Klopfleisch, R.; Loh, G.; Blaut, M. Commensal Akkermansia muciniphila exacerbates gut inflammation in Salmonella Typhimurium-infected gnotobiotic mice. PLoS ONE 2013, 8, e74963. [Google Scholar] [CrossRef]
  45. Chia, L.W.; Hornung, B.V.H.; Aalvink, S.; Schaap, P.J.; de Vos, W.M.; Knol, J.; Belzer, C. Deciphering the trophic interaction between Akkermansia muciniphila and the butyrogenic gut commensal Anaerostipes caccae using a metatranscriptomic approach. Antonie Van Leeuwenhoek 2018, 111, 859–873. [Google Scholar] [CrossRef] [PubMed]
  46. Shuoker, B.; Pichler, M.J.; Jin, C.; Sakanaka, H.; Wu, H.; Gascuena, A.M.; Liu, J.; Nielsen, T.S.; Holgersson, J.; Nordberg Karlsson, E.; et al. Sialidases and fucosidases of Akkermansia muciniphila are crucial for growth on mucin and nutrient sharing with mucus-associated gut bacteria. Nat. Commun. 2023, 14, 1833. [Google Scholar] [CrossRef]
  47. Wang, B.; Chen, X.; Chen, Z.; Xiao, H.; Dong, J.; Li, Y.; Zeng, X.; Liu, J.; Wan, G.; Fan, S.; et al. Stable colonization of Akkermansia muciniphila educates host intestinal microecology and immunity to battle against inflammatory intestinal diseases. Exp. Mol. Med. 2023, 55, 55–68. [Google Scholar] [CrossRef]
  48. Chen, T.; Wang, R.; Duan, Z.; Yuan, X.; Ding, Y.; Feng, Z.; Bu, F.; Liu, L.; Wang, Q.; Zhou, J.; et al. Akkermansia muciniphila Protects Against Psychological Disorder-Induced Gut Microbiota-Mediated Colonic Mucosal Barrier Damage and Aggravation of Colitis. Front. Cell Infect. Microbiol. 2021, 11, 723856. [Google Scholar] [CrossRef]
  49. Boutagy, N.E.; McMillan, R.P.; Frisard, M.I.; Hulver, M.W. Metabolic endotoxemia with obesity: Is it real and is it relevant? Biochimie 2016, 124, 11–20. [Google Scholar] [CrossRef]
  50. Nian, F.; Wu, L.; Xia, Q.; Tian, P.; Ding, C.; Lu, X. Akkermansia muciniphila and Bifidobacterium bifidum Prevent NAFLD by Regulating FXR Expression and Gut Microbiota. J. Clin. Transl. Hepatol. 2023, 11, 763–776. [Google Scholar] [CrossRef]
  51. Ottman, N.; Geerlings, S.Y.; Aalvink, S.; de Vos, W.M.; Belzer, C. Action and function of Akkermansia muciniphila in microbiome ecology, health and disease. Best. Pract. Res. Clin. Gastroenterol. 2017, 31, 637–642. [Google Scholar] [CrossRef]
  52. Kozlovski, S.; Regev, O.; Sapoznikov, A.; Kizner, M.; Achdout, H.; Petrovich-Kopitman, E.; Elkahal, J.; Addadi, Y.; Silva Castanheira, F.V.E.; Feigelson, S.W.; et al. ICAMs are dispensable for influenza clearance and anti-viral humoral and cellular immunity. Front. Immunol. 2022, 13, 1041552. [Google Scholar] [CrossRef]
  53. Bonnechere, B.; Amin, N.; van Duijn, C. What Are the Key Gut Microbiota Involved in Neurological Diseases? A Systematic Review. Int. J. Mol. Sci. 2022, 23, 13665. [Google Scholar] [CrossRef] [PubMed]
  54. Cekanaviciute, E.; Yoo, B.B.; Runia, T.F.; Debelius, J.W.; Singh, S.; Nelson, C.A.; Kanner, R.; Bencosme, Y.; Lee, Y.K.; Hauser, S.L.; et al. Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models. Proc. Natl. Acad. Sci. USA 2017, 114, 10713–10718. [Google Scholar] [CrossRef] [PubMed]
  55. Berer, K.; Gerdes, L.A.; Cekanaviciute, E.; Jia, X.; Xiao, L.; Xia, Z.; Liu, C.; Klotz, L.; Stauffer, U.; Baranzini, S.E.; et al. Gut microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalomyelitis in mice. Proc. Natl. Acad. Sci. USA 2017, 114, 10719–10724. [Google Scholar] [CrossRef]
  56. Li, X.; Lin, D.; Hu, X.; Shi, X.; Huang, W.; Ouyang, Y.; Chen, X.; Xiong, Y.; Wu, X.; Hong, D.; et al. Akkermansia muciniphila Modulates Central Nervous System Autoimmune Response and Cognitive Impairment by Inhibiting Hippocampal NLRP3-Mediated Neuroinflammation. CNS Neurosci. Ther. 2025, 31, e70320. [Google Scholar] [CrossRef]
  57. Steimle, A.; Neumann, M.; Grant, E.T.; Willieme, S.; De Sciscio, A.; Parrish, A.; Ollert, M.; Miyauchi, E.; Soga, T.; Fukuda, S.; et al. Gut microbial factors predict disease severity in a mouse model of multiple sclerosis. Nat. Microbiol. 2024, 9, 2244–2261. [Google Scholar] [CrossRef]
  58. Targher, G.; Valenti, L.; Byrne, C.D. Metabolic Dysfunction-Associated Steatotic Liver Disease. N. Engl. J. Med. 2025, 393, 683–698. [Google Scholar] [CrossRef]
  59. Kim, S.; Lee, Y.; Kim, Y.; Seo, Y.; Lee, H.; Ha, J.; Lee, J.; Choi, Y.; Oh, H.; Yoon, Y. Akkermansia muciniphila Prevents Fatty Liver Disease, Decreases Serum Triglycerides, and Maintains Gut Homeostasis. Appl. Environ. Microbiol. 2020, 86, e03004-19. [Google Scholar] [CrossRef]
  60. Keshavarz Azizi Raftar, S.; Ashrafian, F.; Yadegar, A.; Lari, A.; Moradi, H.R.; Shahriary, A.; Azimirad, M.; Alavifard, H.; Mohsenifar, Z.; Davari, M.; et al. The Protective Effects of Live and Pasteurized Akkermansia muciniphila and Its Extracellular Vesicles against HFD/CCl4-Induced Liver Injury. Microbiol. Spectr. 2021, 9, e0048421. [Google Scholar] [CrossRef]
  61. Oguri, N.; Miyoshi, J.; Nishinarita, Y.; Wada, H.; Nemoto, N.; Hibi, N.; Kawamura, N.; Miyoshi, S.; Lee, S.T.M.; Matsuura, M.; et al. Akkermansia muciniphila in the small intestine improves liver fibrosis in a murine liver cirrhosis model. NPJ Biofilms Microbiomes 2024, 10, 81. [Google Scholar] [CrossRef]
  62. Lakshmanan, A.P.; Murugesan, S.; Al Khodor, S.; Terranegra, A. The potential impact of a probiotic: Akkermansia muciniphila in the regulation of blood pressure-the current facts and evidence. J. Transl. Med. 2022, 20, 430. [Google Scholar] [CrossRef]
  63. Nogal, A.; Valdes, A.M.; Menni, C. The role of short-chain fatty acids in the interplay between gut microbiota and diet in cardio-metabolic health. Gut Microbes 2021, 13, 1897212. [Google Scholar] [CrossRef]
  64. Psichas, A.; Sleeth, M.L.; Murphy, K.G.; Brooks, L.; Bewick, G.A.; Hanyaloglu, A.C.; Ghatei, M.A.; Bloom, S.R.; Frost, G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. 2015, 39, 424–429. [Google Scholar] [CrossRef]
  65. Byrne, C.S.; Chambers, E.S.; Morrison, D.J.; Frost, G. The role of short chain fatty acids in appetite regulation and energy homeostasis. Int. J. Obes. 2015, 39, 1331–1338. [Google Scholar] [CrossRef]
  66. Zhang, D.; Jian, Y.P.; Zhang, Y.N.; Li, Y.; Gu, L.T.; Sun, H.H.; Liu, M.D.; Zhou, H.L.; Wang, Y.S.; Xu, Z.X. Short-chain fatty acids in diseases. Cell Commun. Signal 2023, 21, 212. [Google Scholar] [CrossRef]
  67. Bae, M.; Cassilly, C.D.; Liu, X.; Park, S.M.; Tusi, B.K.; Chen, X.; Kwon, J.; Filipcik, P.; Bolze, A.S.; Liu, Z.; et al. Akkermansia muciniphila phospholipid induces homeostatic immune responses. Nature 2022, 608, 168–173. [Google Scholar] [CrossRef] [PubMed]
  68. Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef] [PubMed]
  69. Ndumele, C.E.; Rangaswami, J.; Chow, S.L.; Neeland, I.J.; Tuttle, K.R.; Khan, S.S.; Coresh, J.; Mathew, R.O.; Baker-Smith, C.M.; Carnethon, M.R.; et al. Cardiovascular-Kidney-Metabolic Health: A Presidential Advisory From the American Heart Association. Circulation 2023, 148, 1606–1635. [Google Scholar] [CrossRef] [PubMed]
  70. de la Cuesta-Zuluaga, J.; Mueller, N.T.; Corrales-Agudelo, V.; Velasquez-Mejia, E.P.; Carmona, J.A.; Abad, J.M.; Escobar, J.S. Metformin Is Associated With Higher Relative Abundance of Mucin-Degrading Akkermansia muciniphila and Several Short-Chain Fatty Acid-Producing Microbiota in the Gut. Diabetes Care 2017, 40, 54–62. [Google Scholar] [CrossRef]
  71. Pellegrino, A.; Coppola, G.; Santopaolo, F.; Gasbarrini, A.; Ponziani, F.R. Role of Akkermansia in Human Diseases: From Causation to Therapeutic Properties. Nutrients 2023, 15, 1815. [Google Scholar] [CrossRef]
Table 1. Human clinical studies of Akkermansia muciniphila (or consortia including A. muciniphila).
Table 1. Human clinical studies of Akkermansia muciniphila (or consortia including A. muciniphila).
Study (Year)Design & PopulationIntervention (Dose; Formulation)ControlDurationPrimary/Key OutcomesSafety/TolerabilityLimitations
Depommier et al., 2019 [18]RCT, double-blind, single-center; overweight/obese insulin-resistant adults; n = 40 randomized (32 completed)Live A. muciniphila 1010/day or pasteurized A. muciniphila 1010/day (oral)Placebo3 monthsPasteurized arm: ↑ insulin sensitivity +28.6% (p = 0.002), ↓ insulinemia −34.1% (p = 0.006), ↓ total cholesterol −8.7% (p = 0.02); weight/fat mass trends only (NS)Well tolerated; AE rates similar to placeboSmall, single-center; modest n, 3-month exposure; not powered for weight loss; multiple endpoints; insulin-resistant but not T2D—limits generalizability.
Zhang et al., 2025 (AKK-WST01) [26]Phase 2 RCT, double-blind, placebo-controlled; drug-naïve adults with overweight/obese T2DM; n = 58Live A. muciniphila (AKK-WST01), daily (oral)Placebo12 weeksOverall: weight & HbA1c fell similarly in both arms (no between-group difference); pre-specified subgroup with low baseline Akkermansia: successful colonization + significant ↓ body weight, ↓ fat mass (incl. visceral), ↓ HbA1cWell toleratedShort duration; modest n; lifestyle counseling in both arms; efficacy driven by baseline-defined subgroup (risk of over-interpretation); single-country—external validity considerations.
Perraudeau et al., 2020 (WBF-011) [28]Multicenter RCT, double-blind, placebo-controlled; adults with T2D on standard care; ITT n = 76Five-strain consortium including A. muciniphila + Anaerobutyricum hallii twice daily (oral capsules)Placebo (and WBF-010 3-strain backbone arm)12 weeksvs placebo: ↓ post-prandial glucose AUC −36.1 mg/dL·180 min (p = 0.0500); ΔHbA1c −0.6% (p = 0.054); within-group −32.5% total post-prandial glucoseWell tolerated; no major safety signalsConsortium design—cannot attribute effects to Akkermansia specifically; borderline p value for primary; modest n; 12-week exposure; sponsor involvement.
Aalipanah et al., 2025 [27]RCT, double-blind; adults with overweight/obesity; n = 66Yogurt with heat-inactivated A. muciniphila (“postbiotic” food)Control yogurt; comparator yogurt with L. rhamnosus postbiotic8 weeks↓ waist circumference, ↓ waist-to-height ratio, ↓ body fat %, ↓ AST vs. control; comparator L. rhamnosus yogurt: no between-group benefitsWell toleratedShort 8-week duration; small sample; food matrix (yogurt) may introduce dietary/placebo-taste effects; anthropometric and enzyme surrogates—not hard clinical endpoints.
AE, adverse event; AUC, area under the curve; HbA1c, glycated hemoglobin; NS, not significant; RCT, randomized controlled trial; T2D, type 2 diabetes. ↑ indicates increase; ↓ indicates decrease.
Table 2. Disease models and context-dependent effects (preclinical).
Table 2. Disease models and context-dependent effects (preclinical).
Domain/ModelPreparationDirection of EffectEndpointsNotes/Caveats
Diet-induced obesity (mice)Live/pasteurized cells; EVsBeneficial↓ weight gain/fat mass; ↑ insulin sensitivity; ↓ adipose inflammationEarly work suggested live > HK; later pasteurized also effective via surface proteins
MASLD/steatosis (mice)Live cells (±antibiotics pretreat)Beneficial↓ liver fat; ↓ TNF-α/IL-6; tissue-specific FXR changesAnti-fibrotic signals model-dependent; not uniform across MASH models
Atherosclerosis (Apoe−/− mice)Live cellsBeneficial↓ lesion area/size; ↓ aortic MCP-1/ICAM-1/TNF-α; ↓ LPSProtection mediated via reduced endotoxemia; HK ineffective in this model
DSS colitis (mice)Cells/Amuc_1100/Amuc_2109Beneficial↓ clinical/histologic colitis; ↓ TNF-α/IL-1β; ↑ junction proteinsMultiple components reproduce benefit
IL-10−/− colitis (mice)CellsPotentially harmful↑ colitis severity/onsetSevere barrier compromise context; timing matters
Acute Salmonella infection (gnotobiotic mice)CellsPotentially harmful↑ inflammationInfection model caveat
DSS, Dextran sulfate sodium; EV, Extracellular vesicle; FXR, Farnesoid X receptor; HK, heat killed; ICAM, Intercellular adhesion molecule; IL, Interleukin; LPS, Lipopolysaccharide; MASH, Metabolic dysfunction-associated steatohepatitis; MASLD, Metabolic dysfunction-associated steatotic liver disease; MCP, Monocyte chemoattractant protein; TNF, Tumor necrosis factor; ↑ indicates increase; ↓ indicates decrease.
Table 3. Practical use considerations.
Table 3. Practical use considerations.
ConsiderationRationaleSuggested Approach in Practice
Baseline Akkermansia abundanceEfficacy of live strain may depend on low baseline levels (colonization succeeds)Consider baseline microbiome profiling for precision use
Formulation (live vs. pasteurized)Pasteurized showed clear metabolic signals in RCT; safer/logistically easierFavor pasteurized in general wellness; reserve live for trials/defined use cases
Special populations (IBD flares, severe barrier defects, certain neurologic conditions)Context-dependent effects; some models show harmAvoid during active severe mucosal injury; consider remission-maintenance trials
Combination strategiesMetformin enriches Akkermansia; GLP-1R agonists shift microbiomeStudy microbe–drug and synbiotic combinations prospectively
Outcomes & durationCurrent human trials are short; hard outcomes pendingDesign/seek longer RCTs with weight/A1c and cardiometabolic endpoints
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

FakhriRavari, A.; Nguyen, M.H.C. Akkermansia muciniphila in Cardiometabolic Medicine: Mechanisms, Clinical Studies, and Therapeutic Outlook. Gastrointest. Disord. 2025, 7, 72. https://doi.org/10.3390/gidisord7040072

AMA Style

FakhriRavari A, Nguyen MHC. Akkermansia muciniphila in Cardiometabolic Medicine: Mechanisms, Clinical Studies, and Therapeutic Outlook. Gastrointestinal Disorders. 2025; 7(4):72. https://doi.org/10.3390/gidisord7040072

Chicago/Turabian Style

FakhriRavari, Alireza, and Minh Hien Chau Nguyen. 2025. "Akkermansia muciniphila in Cardiometabolic Medicine: Mechanisms, Clinical Studies, and Therapeutic Outlook" Gastrointestinal Disorders 7, no. 4: 72. https://doi.org/10.3390/gidisord7040072

APA Style

FakhriRavari, A., & Nguyen, M. H. C. (2025). Akkermansia muciniphila in Cardiometabolic Medicine: Mechanisms, Clinical Studies, and Therapeutic Outlook. Gastrointestinal Disorders, 7(4), 72. https://doi.org/10.3390/gidisord7040072

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop